fas mrna
Recently Published Documents


TOTAL DOCUMENTS

66
(FIVE YEARS 7)

H-INDEX

19
(FIVE YEARS 0)

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3492-3492
Author(s):  
Martina Antoniolli ◽  
Maria Solovey ◽  
Deepak Bararia ◽  
Carolin Dorothea Strobl ◽  
William David Keay ◽  
...  

Abstract Follicular lymphoma (FL) is a clinically and genetically heterogeneous disease. Somatic gene mutations contribute to the heterogeneous clinical course of FL. ARID1A, which encodes for a subunit of the SWI/SNF chromatin remodeling complex, is among the most commonly mutated genes in FL (up to 15% of cases). These mutations are mostly disruptive and are predicted to result in protein haplodeficiency. While we have previously shown that ARID1A mutations are predictive of treatment outcome (Pastore, 2015), the underlying biology of ARID1A loss in FL is unclear. A functional genome-wide in vitro screen showed that ARID1A loss rescued a number of cancer cell lines from FAS-L induced apoptosis (Luo, 2008). FAS-L induced apoptosis plays a critical role in normal B-cell development and homeostasis. Thus, FAS/FAS-L deficiency could contribute to FL development and disease biology. Therefore, we studied the role of ARID1A loss in FAS expression and regulation. We first tested FAS-L induced apoptosis in established lymphoma cell lines that harbor the FL-hallmark translocation t(14;18)[BCL2/IGH] plus ARID1A mutations (Karpas422, WSU-FSCCL) or no ARID1A mutations (OCI-Ly1, OCI-Ly8, SU-DHL16). ARID1A mutant (mut) cells were indeed markedly less sensitive to FAS-L (300 ng/mL/24 hrs) compared to ARID1A wild type (WT) cells (98% vs 52% mean viability by Annexin-V). FAS receptor expression on mutant cells was reduced by almost half compared to WT cells by FACS analysis (N=3, P=0.0004). To test if reduced FAS expression was directly linked to ARID1A loss, we generated single-cell derived clones (from OCI-Ly1 and OCI-Ly8) with either heterozygous (het) loss or complete knock-out (KO) of ARID1A by CRISPR/Cas9. ARID1A loss was validated by Sanger sequencing and Western blot. We consistently observed significantly reduced FAS-L induced apoptosis in het and KO clones (exemplary shown for OCI-Ly8 in Fig A). Remarkably, re-expressed of ARID1A in het cells (het+ARID1A) rescued sensitivity to FAS-L induced apoptosis (Fig A). We confirmed reduced FAS expression on mutant clones by FACS, while re-expression of ARID1A rescued its expression (Fig B). Furthermore, FAS mRNA expression was significantly reduced by qPCR in mut vs WT clones (N=4, P<0.05), while FAS mRNA levels were rescued to WT levels in het+ARID1A cells. To understand the molecular mechanism that links ARID1A loss and reduced FAS expression, we performed ATAC sequencing (Seq) and RNA Seq on 15 single-cell derived clones (9 mut and 6 WT from OCI-Ly1 and OCI-Ly8). RNA Seq confirmed significantly lower ARID1A and FAS mRNA levels (adj p<0.001 each) in the mut clones. We first hypothesized that ARID1A loss could directly affect chromatin accessibility at the FAS promoter. However, we did not observe different chromatin accessibility at the FAS promoter. Next, we searched our data for all known FAS-regulating transcription factors (TFs) (https://dorothea.opentargets.io/#/), but could not identify candidates that were both differentially accessible and differentially expressed. Finally, we searched our data for transcriptional networks, i.e. hubs of all recognized FAS-regulating TFs and their known and predicted interacting partners (https://string-db.org/). Through this, we identified RUNX3, a predicted Co-TF of ETS1, to be both less accessible ("closed chromatin") and less expressed upon ARID1A loss (Fig C), suggesting a novel ARID1A-dependent FAS-regulatory network. To functionally validate our model, we first confirmed reduced RUNX3 expression in ARID1A mutant clones by qPCR and Western blot, and showed that ETS1 levels were unaffected by ARID1A loss. Then, we stably overexpressed RUNX3 in ARID1A mutant clones by lentiviral transduction and could indeed show rescue of FAS surface levels by FACS (Fig D). Lastly, we wanted to validate our findings in primary patients samples. We quantified FAS expression in FL biopsies with known ARID1A mutation status by nCounter gene expression profiling (GEP; N=51, 12 mut vs 39 WT) and quantitative multispectral imaging (QMI; N=44, 10 mut vs 34 WT) (Fig E). Both approaches showed significantly reduced FAS expression in ARID1A mutant FL (P<0.05 for GEP, P<0.0001 for QMI; Fig E). In summary, we show that ARID1A loss is directly linked to reduced FAS expression via a novel RUNX3/ETS1 transcriptional network, potentially opening avenues for therapeutic targeting of this clinically relevant perturbation. Figure 1 Figure 1. Disclosures Subklewe: Pfizer: Consultancy, Speakers Bureau; Takeda: Speakers Bureau; Klinikum der Universität München: Current Employment; Janssen: Consultancy; Seattle Genetics: Consultancy, Research Funding; Roche: Research Funding; Novartis: Consultancy, Research Funding, Speakers Bureau; MorphoSys: Research Funding; Miltenyi: Research Funding; Gilead: Consultancy, Research Funding, Speakers Bureau; Amgen: Consultancy, Research Funding, Speakers Bureau; BMS/Celgene: Consultancy, Research Funding, Speakers Bureau. von Bergwelt: Kite/Gilead: Honoraria, Research Funding, Speakers Bureau; Roche: Honoraria, Research Funding, Speakers Bureau; Novartis: Honoraria, Research Funding, Speakers Bureau; Astellas: Honoraria, Research Funding, Speakers Bureau; Miltenyi: Honoraria, Research Funding, Speakers Bureau; BMS: Honoraria, Research Funding, Speakers Bureau; Mologen: Honoraria, Research Funding, Speakers Bureau; MSD Sharpe & Dohme: Honoraria, Research Funding, Speakers Bureau. Weigert: Janssen: Speakers Bureau; Epizyme: Membership on an entity's Board of Directors or advisory committees; Roche: Research Funding.


Nutrients ◽  
2021 ◽  
Vol 13 (8) ◽  
pp. 2501
Author(s):  
Maihemuti Mijiti ◽  
Ryosuke Mori ◽  
Bingyu Huang ◽  
Kenichiro Tsukamoto ◽  
Keisuke Kiriyama ◽  
...  

Dietary protamine can ameliorate hyperlipidemia; however, the protamine-derived active peptide and its hypolipidemic mechanism of action are unclear. Here, we report the discovery of a novel anti-obesity and hypocholesterolemic peptide, RPR (Arg-Pro-Arg), derived from protamine in mice fed a high-fat diet for 50 days. Serum cholesterol levels were significantly lower in the protamine and RPR groups than in the control group. White adipose tissue weight was significantly decreased in the protamine and RPR groups. The fecal excretion of cholesterol and bile acid was significantly higher in the protamine and RPR groups than in the control group. We also observed a significant decrease in the expression of hepatic SCD1, SREBP1, and adipocyte FAS mRNA, and significantly increased expression of hepatic PPARα and adipocyte PPARγ1 mRNA in the protamine group. These findings demonstrate that the anti-obesity effects of protamine are linked to the upregulation of adipocyte PPARγ1 and hepatic PPARα and the downregulation of hepatic SCD1 via SREBP1 and adipocyte FAS. RPR derived from protamine has a crucial role in the anti-obesity action of protamine by evaluating the effective dose of adipose tissue weight loss.


2021 ◽  
pp. 1-13
Author(s):  
Yi Zhang ◽  
Xuan Shao ◽  
Chenyi Gao ◽  
Danying Xu ◽  
Jun Wu ◽  
...  

BACKGROUND: FAS can serve as both an oncogene and a suppresser in different malignancies, and the prognostic value of FAS remains controversial. METHODS: The Oncomine database, KM-Plotter and bc-GenExMiner platform were adopted to analyze the prognostic value of FAS in breast cancer. Breast cancer tissue microarrays were further used to verify these data. The Cell Miner Tool was used to predict the value of FAS mRNA expression in predicting the efficacies of clinical drugs. RESULTS: We found that both FAS mRNA and protein expression level significantly reduced in breast carcinoma. In addition, high FAS expression indicates a better metastatic relapse-free survival. Interestingly, FAS was associated with a better prognosis in different subtypes of breast cancer patients, namely, only in grade II and III, lymph nodal positive or p53 wild-type patients. The data from the Cell Miner Tool revealed that FAS mRNA expression was correlated with the efficacy of the first-line chemotherapeutic taxane agents and target drugs including olaparib and everolimus. CONCLUSIONS: FAS expression correlates with a better prognosis in breast cancer and may provide an effective clinical strategy to predict the sensitivity of taxanes and targeted drugs.


2021 ◽  
Vol 22 (7) ◽  
pp. 3631
Author(s):  
Hsiao-Chien Lin ◽  
Jin-Yuarn Lin

The inhibitory effects of purified fractions isolated from guava seed polysaccharides (GSPS) including guava seed polysaccharide fraction 1 (GSF1), GSF2, and GSF3 on prostate cancer cells remain unclear. To clarify the anti-prostate cancer potential, GSPS, GSF1, GSF2, and GSF3 were isolated using Sepharose 6B gel filtration chromatography to assay their inhibitory effects on prostate PC-3 cell growth with direct action or indirect immunotherapy using either splenocyte conditioned media (SCM) or macrophage conditioned media (MCM). Correlations between cytokine profiles in the conditioned media and pro-apoptotic gene expression levels in the corresponding treated PC-3 cells were analyzed. Results showed that GSPS, GSF1, GSF2, and GSF3, particularly GSF3, through either direct action or indirect treatments using SCM or MCM, significantly (p < 0.05) inhibited PC-3 cell growth. GSF3 direct treatments increased pro-apoptotic Bax/anti-apoptotic Bcl-2 mRNA expression ratios in corresponding treated PC-3 cells. Either SCM or MCM cultured with GSF3 increased Fas mRNA expression levels in corresponding treated PC-3 cells. Both Th2-polarized and anti-inflammatory cytokine IL-10 either secreted in SCM or MCM were positively correlated with Fas mRNA expression levels in corresponding treated PC-3 cells. Our results suggest that GSF3 is a potent biological response modifier to decrease PC-3 cell growth through inducing apoptosis.


2020 ◽  
Vol 11 (10) ◽  
Author(s):  
Tianke Chen ◽  
Xiaowei Dai ◽  
Juji Dai ◽  
Chaodong Ding ◽  
Zheng Zhang ◽  
...  

Abstract Hepatocellular carcinoma (HCC) is a major leading cause of cancer-related death worldwide. Alpha fetoprotein (AFP) is reactivated in a majority of hepatocellular carcinoma (HCC) and associated with poor patient outcomes. Although increasing evidence has shown that AFP can regulate HCC cell growth, the precise functions of AFP in hepatocarcinogenesis and the associated underlying mechanism remain incompletely understood. In this study, we demostrated that depleting AFP significantly suppressed diethylnitrosamine (DEN)-induced liver tumor progression in an AFP gene-deficient mouse model. Similarly, knocking down AFP expression inhibited human HCC cell proliferation and tumor growth by inducing apoptosis. AFP expression level was inversely associated with the apoptotic rate in mouse and human HCC specimens. Investigation of potential cross-talk between AFP and apoptotic signaling revealed that AFP exerted its growth-promoting effect by suppressing the Fas/FADD-mediated extrinsic apoptotic pathway. Mechanistically, AFP bound to the RNA-binding protein HuR, increasing the accumulation of HuR in the cytoplasm and subsequent inhibition of Fas mRNA translation. In addition, we found that inhibiting AFP enhanced the cytotoxicity of therapeutics to AFP-positive HCC cells by activating HuR-mediated Fas/FADD apoptotic signaling. Conclusion: Our study defined the pro-oncogenic role of AFP in HCC progression and uncovered a novel antiapoptotic mechanism connecting AFP to HuR-mediated Fas translation. Our findings suggest that AFP is involved in the pathogenesis and chemosensitivity of HCC and that blockade of AFP may be a promising strategy to treat advanced HCC.


2020 ◽  
Vol 11 (3) ◽  
pp. 542-550
Author(s):  
Zhigang Chen ◽  
Jun Wu ◽  
Hailin Xu ◽  
Xiuyan Yu ◽  
Ke Wang

2018 ◽  
Vol 45 (10) ◽  
pp. 1397-1405 ◽  
Author(s):  
Lin Xie ◽  
Jinhua Xu

Objective.T-lymphocyte apoptosis plays a critical role in the pathogenesis of systemic lupus erythematosus (SLE). However, the underlying regulatory mechanisms of apoptosis in SLE remain unclear. The aim of this study was to explore the role of miR-98 in SLE and its underlying mechanisms.Methods.Western blotting and quantitative reverse transcription PCR (qRT-PCR) were used to analyze miR-98 and Fas expression. Luciferase reporter assays were performed to identify miR-98 targets. To modify miRNA levels, miR-98 mimics and inhibitor were transfected into cells. A lentiviral construct was used to overexpress the level of Fas in SLE CD4+ T cells. Gene and protein expression were determined by qRT-PCR and Western blotting. Apoptosis levels were evaluated by annexin V staining and flow cytometry.Results.Compared to those of healthy donors, miR-98 was downregulated in SLE CD4+ T cells, whereas Fas mRNA and protein expression were upregulated. Upregulation of miR-98 by mimic transfection protected Jurkat cells against Fas-mediated apoptosis at both mRNA and protein levels, while miR-98 inhibitor induced the completely opposite effect. Luciferase reporter assays demonstrated that miR-98 directly targeted Fas mRNA. Further, miR-98 inhibitor induced apoptosis in primary healthy CD4+ T cells through the Fas-caspase axis, while upregulation of miR-98 in SLE CD4+ T cells led to the opposite effect.Conclusion.The current study revealed that downregulation of miR-98 induces apoptosis by modulating the Fas-mediated apoptotic signaling pathway in SLE CD4+ T cells. These results suggest that miR-98 might serve as a potential target for SLE treatment.


2018 ◽  
Vol 21 (02) ◽  
pp. 43-48 ◽  
Author(s):  
Erkigul B ◽  
Gerelt B ◽  
Damdinsuren L

This survey’s aim was to investigate the seasonal effect of Fatty acid synthesis (FAS) expression on intramuscular fat (IMF) content in grazing Mongolian, Ujumqin and grain-fed Ujumqin sheep. Different skeletal muscles of sheep (biceps femoris, longissimus dorsi and triceps brachii) were sampled to measure IMF content and total RNA was extracted to determine FAS mRNA expression levels by real-time PCR. The result revealed that: (1) The difference in IMF content in the muscles of Mongolian sheep grazed in summer was observed to be much higher than sheep grazed in winter (p<0.01). Also there was significant difference in IMF content in the muscles of Ujumqin sheep grazed in summer than winter (p<0.05). The mRNA expression level of FAS in muscles of winter grazing Mongolian sheep was significantly higher than summer (p<0.05). The seasonal effects on IMF content in the muscles and it’s FAS mRNA gene expression was all negative correlated between the sheep breeds. Accordingly, grazing Mongolian sheep’s FAS gene expression level was very high negatively correlated (r=-0.964). This study suggested that grazing Mongolian and Ujumqin sheep able to store large amount of IMF under depends on seasonal effects.


2016 ◽  
Vol 15 (3) ◽  
pp. 34-39 ◽  
Author(s):  
D. A. Afanasieva ◽  
V. A. Misyurin ◽  
A. V. Ponomarev ◽  
I. M. Luchenko ◽  
O. S. Burova ◽  
...  

Background. Aranose is alkylating antineoplastic agent with cytotoxic and cytostatic properties. Liposomal form of aranose has advantages compared with traditional dosage form. Another alkylating agent, cisplatin, promotes mRNA increase expression level of genes, which control external apoptosis way. Objective. We assume that liposomal form of aranose can change the CD95/Fas, TNFR1, TNFR2, TRAIL, DR3 and DR4/5 expression level in melanoma cell lines mel Kor, mel Z, mel Mtp, mel Mtp-X, mel Ibr, mel Ch, mel Is and mel R. Materials and methods. There are two forms of aranose were used in this work: liposomal form and traditional diluted form for injection. Melanoma cell lines were incubated with both forms. We used RQ PCR to evaluate mRNA expression mRNA expression of DR3, DR4/5, CD95/Fas, TNFR1, TNFR2 and TRAIL relatively gene ABL. Results. CD95/FAS mRNA expression level was most changeable. Liposomal aranose increase CD95/FAS expression level in mel Ibr and mel Mtp-X cell lines and reduced CD95/FAS expression level in mel Ch and mel Is. Conclusion. Thus, liposomal aranose can change mRNA CD95/FAS expression level in melanoma cell lines. Using this process, liposomal form can affect the apoptotic cell death of melanoma.


Sign in / Sign up

Export Citation Format

Share Document